Abstract
Long non-coding RNAs (lncRNAs) are a recently discovered class of non-coding RNAs with a length of more than 200 nucleotides. Dysregulation of lncRNAs has been implicated in the development and progression of various human diseases and has pivotal functions in diverse biological processes, like cell apoptosis, proliferation, migration, and invasion. LncRNAs are increasingly being identified as potential targets for clinical applications based on their functional mechanisms, which are gradually being elucidated. One such disease-related lncRNA is the forkhead box P4-AS1 (FOXP4-AS1), which is abnormally expressed in multiple human diseases, including cancer and non-cancerous diseases. Moreover, FOXP4-AS1’s expression is linked with the clinical characteristics of patients. Mechanistically, FOXP4-AS1 mediates several cellular processes mainly through its target genes. Therefore, FOXP4-AS1 has been extensively investigated for its diagnostic, prognostic, and therapeutic potential. In this review, we discuss the expression patterns, major role, and molecular mechanisms of FOXP4-AS1, along with its association with clinical features and potential applications in clinical settings.
Introduction
With the continuous development of medical technologies and the advancement of research, genomic medical science and its relationship with the causes of human disorders have received growing attention [1], [2], [3], [4], [5], [6]. Given the widespread application of high-throughput RNA sequencing technologies long non-coding RNAs (lncRNAs), a class of non-coding RNAs with a length of more than 200 nucleotides were recently discovered [7], [8], [9], [10], [11]. LncSEA [12] is a platform with the largest number of human lncRNA sets, having comprehensive annotation with information on enrichment analysis and associated upstream regulators and downstream targets of lncRNAs. The powerful lncRNA platform supports researchers to obtain highly specific functions of lncRNAs of interest. Numerous studies have uncovered that lncRNAs perform significant functions in various physiological and pathological processes, including cell cycle, proliferation, differentiation, apoptosis, inflammatory response, and invasion [13], [14], [15], [16], [17]. LncRNAs regulate gene expression and function through interactions with diverse molecules, such as proteins, RNA and DNA [18], [19], [20], [21], [22], [23], [24]. These unique signatures suggest the crucial advantages of lncRNAs for their diagnostic, prognostic, and therapeutic roles in diverse human disorders [25], [26], [27], [28].
One disease-specific lncRNA of interest is the lncRNA forkhead box P4 (FOXP4)-AS1 (FOXP4-AS1), a recently discovered antisense transcript of FOXP4 located on the chromosome 6p21.1. Its size is 96,181 bases. Several studies suggest that FOXP4-AS1 expression is aberrantly regulated in human diseases, including colorectal cancer [29], [30], [31], esophageal cancer [32, 33], nasopharyngeal cancer [34], [35], [36], liver cancer [37], [38], [39], gastric cancer [40], ovarian cancer [41, 42], cervical cancer [43], lymphoma [44], lung cancer [45], bone cancer [46, 47], pancreatic cancer [48], prostate cancer [49], glioma [50], COVID-19 [51, 52], placenta accreta [53], and thyroid cancer [54]. FOXP4-AS1 levels are strongly correlated with patients’ clinical features and prognosis, along with their TNM stage, tumor size, and lymph node metastasis [32, 34, 47]. FOXP4-AS1 participated extensively in disease progression through the regulation of important cellular processes, like cell apoptosis, proliferation, migration, and invasion [29, 32, 33, 35]. Mechanistic studies have provided a deeper insight into FOXP4-AS1’s interactions with its target genes. FOXP4-AS1 exerts its influence on biological processes by directly modulating the expression of downstream target genes [44, 45]. Functioning as a regulatory factor, it interacts with the DNA or RNA sequence of these target genes, thereby exerting an impact on gene transcription and translational processes. This regulatory mechanism alters the functional state of cells, thereby affecting processes such as cellular proliferation, differentiation, and apoptosis. Furthermore, FOXP4-AS1 is regulated by its upstream molecules. Transcription factors, miRNAs, proteins, and other molecules can interact with FOXP4-AS1, thereby controlling its expressional level within the cell [49]. This upstream regulatory mechanism indirectly affects the expression of downstream target genes by modulating the stability, transcription rate, or translational process of FOXP4-AS1. Consequently, these properties render FOXP4-AS1 as a potential clinical biomarker of disease diagnosis, prognosis as well as a therapeutic target [32, 34, 47, 50].
In this review, we focus on the current understanding of FOXP4-AS1 from the aspects of its expression, functions, and regulatory mechanisms, and discuss its promising clinical applications. Furthermore, recent advancements in the field are reviewed, along with future directions to deepen the understanding of FOXP4-AS1 and its implications in human disorders.
FOXP4-AS1’s expression and its roles in human diseases
The aberrant expression of FOXP4-AS1 plays a significant role in the development and progression of various diseases, including colorectal cancer [29], [30], [31], esophageal cancer [32, 33], nasopharyngeal cancer [34], [35], [36], liver cancer [37], [38], [39], gastric cancer [40], ovarian cancer [41, 42], cervical cancer [43], lymphoma [44], lung cancer [45], bone cancer [46, 47], pancreatic cancer [48], prostate cancer [49], glioma [50], COVID-19 [51, 52], placenta accreta [53], and thyroid cancer [54] (Figure 1). Moreover, there exists a strong correlation between the levels of FOXP4-AS1 expression and clinicopathological features, including overall survival (OS), TNM stage, tumor size, and lymph node metastasis (Table 1). The dysregulation of FOXP4-AS1 has been implicated in the pathogenesis of these diseases through various molecular mechanisms.

The dysregulation of FOXP4-AS1 in human cancers.
LncRNA FOXP4-AS1 expression and clinical characteristics in human diseases.
Disease type | Expression | Clinical characteristics | Refs |
---|---|---|---|
Colorectal cancer | Upregulated | TNM stages, and tumor size | [29], [30], [31] |
Esophageal cancer | Upregulated | Lymph node metastasis, TNM stage, OS, and RFS | [32, 33] |
Nasopharyngeal cancer | Upregulated | T stage, clinical stage, lymph node metastasis, OS, and PFS | [34], [35], [36] |
Liver cancer | Upregulated | Age, AFP, AST, BCLC stage, tumor size, DFS, and OS | [37], [38], [39] |
Gastric cancer | Upregulated | DFS | [40] |
Ovarian cancer | Downregulated | FIGO stage, clinical stage, grades, lymphatic invasion, OS, DFS, and PFS | [41, 42] |
Cervical cancer | Upregulated | – | [43] |
Lymphoma | Upregulated | OS, DFS, and clinical stages | [44] |
Lung cancer | Upregulated | – | [45] |
Ewing Sarcoma | Upregulated | OS, and EFS | [46] |
Osteosarcoma | Upregulated | – | [47] |
Pancreatic cancer | Upregulated | OS, and mortality rates | [48] |
Prostate cancer | Upregulated | OS, and tumor stage | [49] |
Glioma | Upregulated | – | [50] |
Papillary thyroid carcinoma | Downregulated | TNM stages, extraglandular invasion, and disease-free interval rate | [54] |
In this section, we provide a comprehensive discussion of the expression patterns of FOXP4-AS1, its association with clinical factors, and its main major functions in several human disorders.
The abnormal expression of FOXP4-AS1 was observed in colorectal cancer, esophageal cancer, nasopharyngeal cancer, liver cancer, gastric cancer, ovarian cancer, cervical cancer, lymphoma, lung cancer, bone cancer, pancreatic cancer, prostate cancer, and thyroid cancer.
Colorectal cancer
Previous studies demonstrated the high expression of FOXP4-AS1 in colorectal cancer cells, HT29, SW620, SW480, LOVO, DLD1, and HCT116 and tissues [29], [30], [31]. High levels of FOXP4-AS1 are positively associated with worse TNM stages and larger tumor size. Moreover, FOXP4-AS1 promotes cancer by increasing tumor cell proliferation, migration, and invasion and suppressing apoptosis, as evidenced in DLD1, HCT116, and HT29 cells. Moreover, FOXP4-AS1 enhances tumor xenograft growth. The above evidence suggests that FOXP4-AS1 plays a crucial role in the progression and aggressiveness of colorectal cancer. Further understanding of its molecular mechanisms may yield potential therapeutic targets and prognostic indicators for this disease.
Esophageal cancer
FOXP4-AS1 is upregulated in esophageal squamous cell carcinoma (ESCC) tissues and Eca109, TE1, TE13, YES-2, KYSE150, EC9706, KYSE410, and KYSE450 cells [32, 33]. Its expression level is positively correlated with the TNM stage of the disease, lymph node metastasis, OS, and recurrence-free survival (RFS). Additionally, FOXP4-AS1 enhances the proliferation, migration, and invasion abilities of YES-2, KYSE450, and KYSE150 cells, thus promoting the progression of ESCC. These findings suggest that FOXP4-AS1 plays a significant role in the development and aggressiveness of ESCC. Further exploration of its underlying mechanisms may yield potential therapeutic targets and prognostic indicators for ESCC.
Nasopharyngeal cancer
In nasopharyngeal carcinoma, upregulated FOXP4-AS1 expression has been observed in the serum and cell lines including CNE1, HONE1, C666-1, and CNE2 [34], [35], [36]. The relationship between FOXP4-AS1 levels and various clinical features of patients, such as the T stage, clinical stage, lymph node metastasis, OS, and progression-free survival (PFS), has also been investigated. Additionally, the overexpression of FOXP4-AS1 promotes cancer development by enhancing CNE1 cell proliferation, apoptosis, metastasis, and epithelial-mesenchymal transition (EMT). These findings suggest that FOXP4-AS1 may play a critical role in the pathogenesis of nasopharyngeal cancer.
Liver cancer
FOXP4-AS1 is upregulated in various hepatocellular carcinoma (HCC) cell lines, including MHCC97H, HepG2, Hep3B, Huh7, LM3, and SMMC7721, as well as in HCC tissues [37], [38], [39]. FOXP4-AS1’s overexpression is associated with several clinical characteristics of HCC patients, such as age, serum aspartate aminotransferase (AST), alpha-fetoprotein (AFP), BCLC stage, tumor size, disease-free survival (DFS), and OS. Furthermore, FOXP4-AS1 aggravates HCC progression and is involved in promoting cell proliferation, invasion, and migration of Hep3B, Huh7, SMMC-7721, and LM3 cells. It can stimulate angiogenesis in human umbilical vein endothelial cells (HUVECs) and increase the size of HCC tumors in mice. These findings highlight the significance of FOXP4-AS1 in the development and aggressiveness of HCC and suggest its potential as a therapeutic target and prognostic marker for this deadly disease.
Gastric cancer
Elevated FOXP4-AS1 levels have been observed in the gastric cancer cells, AGS, NCI-N87, and MKN-45 and tissues, and its overexpression is associated with poorer DFS [40]. These results underscore the potential oncogenic role of FOXP4-AS1 in gastric cancer and suggest its promising role as a therapeutic target.
Ovarian cancer
Recent studies have revealed that the downregulation of FOXP4-AS1 in ovarian cancer restrains EMT and even mediates immune cell infiltration and inflammatory responses [41, 42]. High FOXP4-AS1 expression correlates positively with favorable International Federation of Gynecology and Obstetrics (FIGO) stage, clinical stage, grades, lymphatic invasion, OS, DFS, and PFS of patients with ovarian cancer.
Other diseases
FOXP4-AS1 is differentially upregulated in cervical cancer and accelerates the malignant behavior of Caski and C33A cells, thus worsening the progression of cervical cancer [43]. Similarly, FOXP4-AS1 accelerates mantle cell lymphoma development by elevating the proliferative and metastatic abilities of the JVM-2 and Z138 cells [44]. High FOXP4-AS1 level correlates with poor OS, DFS, and clinical stages in patients with lymphoma. FOXP4-AS1 is upregulated in non-small cell lung cancer (NSCLC) A549, Calu‐1, and H1299 cells as well as tissues [45]. FOXP4-AS1 was proposed as a cancer‐promoting regulator which enhances the proliferative function of A549 and Calu‐1 cells and xenograft tumor growth in nude mice. As for bone cancer, FOXP4-AS1 was found to be remarkably upregulated in Ewing Sarcoma SK-N-MC, and A673 cell lines, and led to worse OS and event-free survival rates (EFS) in these patients. FOXP4-AS1 resulted in aggressive Ewing Sarcoma by enhancing cell invasion, proliferation, and migration [46]. FOXP4-AS1 was also upregulated in HOS, A673, SK-N-MC, MG63, Saos2, and U2OS osteosarcoma cells. FOXP4-AS1 accelerated the proliferation, migration and inhibited the apoptosis of A673, SK-N-MC, MG63, and U2OS cells, thereby promoting tumor formation [47]. Moreover, high FOXP4‐AS1 expression in pancreatic cancer strongly correlates with poor prognosis and was found to increase cancer‐related mortality rates in these patients [48]. FOXP4‐AS1 is highly expressed in prostate cancer PC-3, DU145, VCaP, and LNCaP cells and tissues [49]. High FOXP4‐AS1 levels reflect poorer OS and tumor stage in patients with prostate cancer. In vivo animal studies and in vitro cytological experiments demonstrate that FOXP4-AS1 exerts a crucial function on prostate cancer progression by facilitating cell proliferation and repressing cell apoptosis. FOXP4-AS1 is downregulated in both papillary thyroid cancer (PTC) tissues and cell lines (K1, BCPAP, and TPC-1). Higher expression levels of FOXP4-AS1 are associated with more favorable patient outcomes, lower TNM stages, and reduced extra glandular invasion. Mechanistically, FOXP4-AS1 exerts its inhibitory effects on PTC progression by negatively regulating FOXP4 expression and suppressing the activity of the Akt signaling pathway [54].
Apart from its involvement in cancer, FOXP4-AS1 functions in other human disorders. Notably, a Japanese genome-wide association study (GWAS) reported that FOXP4-AS1 is related to the initiation and severity of the COVID-19 disease [51]. A COVID-19 Host Genetics Initiative (HGI) analysis also suggested that the common variants of FOXP4-AS1 may regulate patients’ adaptive immunity, and therefore, are closely associated with COVID-19 disease severity [52].
Mechanisms of FOXP4-AS1 action in human disorders
Numerous studies have provided evidence for the role of FOXP4-AS1 dysregulation in driving the progression of various human diseases by mediating diverse cellular processes, including proliferation, apoptosis, migration, and invasion [35], [36], [37]. These findings highlight the diverse and multifaceted roles of FOXP4-AS1 in human diseases, underscoring its potential as a diagnostic marker, prognostic predictor, and therapeutic target. Therefore, in this section, we briefly introduce the predominant mechanisms underlying the biological functions of FOXP4-AS1 in several human disorders.
The irregular regulation of cell proliferation and apoptosis often results in uncontrollable cell growth and eventual cancer formation [55], [56], [57], [58]. Aberrant cell migration and invasion drive tissue metastasis, accounting for over 90 % of cancer-associated deaths [59], [60], [61], [62], [63]. Therefore, an in-depth understanding of the mechanisms of these essential biological processes is becoming an increasingly attractive approach for the development of new treatment types. FOXP4-AS1 has been implicated in the development and progression of different types of human disorders, such as CRC, esophageal cancer, nasopharyngeal cancer, liver cancer, gastric cancer, pancreatic cancer, ovarian cancer, cervical cancer, prostate cancer, lung cancer, lymphoma, Ewing Sarcoma, osteosarcoma, glioma, and even COVID-19 (Table 2). Recent studies have shown that FOXP4-AS1 is activated by ATF3, leading to the acceleration of malignant processes in colorectal cancer HCT116 cells. This occurs through the interaction of FOXP4-AS1 with miR-423-5p, resulting in the upregulation of NACC1 expression [29]. Additionally, FOXP4-AS1 promotes cell migration, invasion, and proliferation in esophageal cancer cell lines, KYSE150 and KYSE450, which is mediated by the MLL2/FOXP4 axis by enhancing β-catenin expression. FOXP4-AS1 also interacts with IGF2BP2 proteins and miRNA-3184-5p to regulate FOXP4 levels in esophageal cancer cells (Figure 2) [32, 33]. In nasopharyngeal cancer, FOXP4-AS1 interferes with miR-423-5p expression, leading to elevated STMN1 levels and the positive regulation of proliferative ability of CNE1 cells [35]. FOXP4-AS1 facilitates metastasis and EMT in C666-1 cells through the miR-136-5p/MAPK1 axis [36]. FOXP4-AS1 increases H3K27me3 levels through the recruitment of EZH2, thus inhibiting ZC3H12D expression in HCC SMMC-7721 and LM3 cells and promoting cell proliferation, invasion, and migration [37]. In cervical cancer, upregulated FOXP4-AS1 boosts malignant transformation processes in C33A and Caski cells by binding with miR-136-5p to enhance CBX4 expression [43]. FOXP4-AS1 interacts with miR-423-5p to upregulate NACC1, thereby strengthening the proliferation, migration, and invasion of mantle cell lymphoma JVM-2 and Z138 cells [44]. The proliferative process of NSCLC A549 and Calu‐1 cells is also enhanced by FOXP4-AS1 through its combination with miR-3184-5p to promote EIF5A expression [45]. Moreover, FOXP4-AS1 increases TMPO levels by binding to miR-298 and enhances the progression of cell proliferation, migration, and invasion in Ewing Sarcoma SK-N-MC and A673 cells [46]. Furthermore, FOXP4-AS1 combines with LSD1 and EZH2 to weaken the levels of LATS1, in turn facilitating the proliferation and migration of OS MG63 and U2OS cells [47]. In prostate cancer, after the activation of FOXP4-AS1 by PAX5, FOXP4-AS1 sponges with miR-3184-5p and increases the FOXP4 level, thus enhancing the proliferative ability of PC-3 cells [49]. These findings suggest that FOXP4-AS1 plays a significant role in various cancers, contributing to the regulation of cell proliferation, migration, invasion, and malignant transformation processes.
LncRNA FOXP4-AS1 roles and regulatory mechanisms in human cancers.
Disease type | Role | Cell lines | Functions | Related mechanisms | Refs |
---|---|---|---|---|---|
Colorectal cancer | Tumor promoter | HT29, SW620, SW480, LOVO, DLD1, and HCT116 | Cell proliferation, apoptosis, migration, and invasion | ATF3, miR-423-5p, and NACC1 | [29], [30], [31] |
esophageal cancer | Tumor promoter | Eca109, TE1, TE13, YES-2, KYSE150, EC9706, KYSE410, and KYSE450 | Cell proliferation, migration, and invasion | MLL2, FOXP4, β-catenin, YY1, IGF2BP2, and miR-3184-5p | [32, 33] |
Nasopharyngeal cancer | Tumor promoter | CNE1, HONE1, C666-1, and CNE2 | Cell proliferation, and apoptosis | miR-423-5p, and STMN1 | [34], [35], [36] |
Liver cancer | Tumor promoter | MHCC97H, HepG2, Hep3B, Huh7, LM3, and SMMC7721 | Cell proliferation, migration, and invasion | H3K27me3, EZH2, and ZC3H12D | [37], [38], [39] |
Gastric cancer | Tumor promoter | MKN45, NCIN87, and AGS | – | – | [40] |
Ovarian cancer | Tumor suppressor | – | Inflammatory response | – | [41, 42] |
Cervical cancer | Tumor promoter | Caski, and C33A | Cell proliferation, migration, and invasion | miR-136-5p, and CBX4 | [43] |
Lymphoma | Tumor promoter | JVM-2, and Z138 | Cell proliferation, migration, and invasion | miR-423-5p, and NACC1 | [44] |
Lung cancer | Tumor promoter | A549, Calu‐1, and H1299 | Cell proliferation | miR-3184-5p, and EIF5A | [45] |
Ewing sarcoma | Tumor promoter | A673, and SK-N-MC | Cell proliferation, migration, and invasion | miR-298, and TMPO | [46] |
Osteosarcoma | Tumor promoter | A673, SK-N-MC, HOS, Saos2, MG63, and U2OS | Cell proliferation, apoptosis, migration, and invasion | LATS1, LSD1, and EZH2 | [47] |
Pancreatic cancer | Tumor promoter | – | – | – | [48] |
Prostate cancer | Tumor promoter | PC-3, DU145, VCaP, and LNCaP | Cell proliferation, and apoptosis | PAX5, miR-3184-5p, and FOXP4 | [49] |
Glioma | Tumor promoter | – | Drug resistance | – | [50] |
Papillary thyroid carcinoma | Tumor suppressor | K1, BCPAP, and TPC-1 | Cell proliferation, apoptosis, and migration | FOXP4, and Akt | [54] |

FOXP4-AS1 enhanced cell proliferation, migration, and invasion in esophageal cancer.
In YES-2 and KYSE150 cells, FOXP4-AS1 combined with MLL2 to upregulate the FOXP4 level and further activated β-catenin expression. In KYSE150 and KYSE450 cells, FOXP4-AS1 interacted with IGF2BP2 protein or miR-3184-5p to enhance FOXP4 expression. Meanwhile, YY1 also elevated the transcription of FOXP4-AS1 and FOXP4.
Clinical application prospects of FOXP4-AS1 in human disease intervention
Although many treatment options have been explored and applied in clinical practice, the global disease burden continues to be heavy, especially that of cancers [64], [65], [66], [67], [68]. Therefore, accurate diagnosis and timely treatment are vital for clinical management and optimizing patient prognosis [69], [70], [71], [72], [73].
Recently, there has been growing interest in the clinical application of FOXP4-AS1 owing to its potential for understanding and managing various diseases. FOXP4-AS1 exhibit differential expression levels in different types of diseases, indicating its involvement in disease mechanisms. FOXP4-AS1 levels are strongly correlated with patients’ clinical characteristics and prognoses. Therefore, FOXP4-AS1 levels have potential diagnostic and prognostic roles in indicating disease progression. To assess the diagnostic accuracy of FOXP4-AS1, researchers frequently employ receiver operating characteristic (ROC) curve analysis. This analysis has confirmed the high diagnostic value of FOXP4-AS1 in distinguishing disease tissues from normal tissues. For example, FOXP4-AS1 possesses a high diagnostic value with an area under the curve (AUC) reaching up to 0.8679 for differentiating ESCC from normal specimens [32]. FOXP4-AS1 expression in nasopharyngeal cancer serum samples has a powerful diagnostic value with an AUC of 0.7974 [34]. In OS, an AUC of 0.851 also suggests the strong diagnostic performance of FOXP4-AS1 [47]. The Kaplan-Meier method also reveals the remarkable association between FOXP4-AS1 expression levels and patients’ clinical features and prognosis across diseases. All of the above results reflect the robust prognostic capability of FOXP4-AS1 in diverse human disorders, such as CRC [30], esophageal cancer [32], nasopharyngeal cancer [34], liver cancer [37], [38], [39], gastric cancer [40], ovarian cancer [41], lymphoma [44], Ewing Sarcoma [46], OS [47], pancreatic cancer [48], and glioma [50]. Furthermore, FOXP4-AS1’s dysregulation has been shown to play a role in modulating crucial biological processes and disease progression by interacting with specific molecules. This discovery opens up possibilities for targeted interventions aimed at manipulating FOXP4-AS1’s expression for therapeutic purposes. Additionally, research has highlighted that FOXP4-AS1 expression can predict the sensitivity of glioma patients to temozolomide treatment, offering potential benefits for personalized medicine [50]. Therefore, FOXP4-AS1 has promising clinical applications in disease diagnosis, prognosis, and potentially even in treatment strategies. The differential expression of FOXP4-AS1 in diseases, its ability to distinguish disease tissues, its correlation with clinical characteristics, and its involvement in disease mechanisms render it an attractive target for further research and development in the field of precision medicine.
In summary, FOXP4-AS1 acts as a disease-related lncRNA and is dysregulated in multiple human diseases, including CRC, esophageal cancer, nasopharyngeal cancer, liver cancer, gastric cancer, ovarian cancer, cervical cancer, lymphoma, lung cancer, bone cancer, pancreatic cancer, prostate cancer, COVID-19, and placenta accreta. Additionally, FOXP4-AS1 levels are closely linked to patient clinical characteristics and prognosis, including age, TNM stage, tumor size, lymph node metastasis, OS, DFS, and RFS. Moreover, FOXP4-AS1 interacts with various target genes, leading to the modulation of crucial cellular processes, like cell invasion, proliferation, migration, apoptosis, and drug sensitivity, all of which suggest the significant role of FOXP4-AS1 in disease progression and treatment response. Given its extensive role and association with human diseases, FOXP4-AS1 is a promising clinical tool for disease diagnosis, prognosis, and designing novel treatment strategies. While the current understanding of FOXP4-AS1 roles is in the initial phase, further in-depth experimental studies and clinical trials are needed to elucidate the underlying biological mechanisms and validate their clinical utility. Looking ahead, future research on FOXP4-AS1 should focus on exploring its specific molecular interactions and downstream signaling pathways. Furthermore, large-scale clinical studies involving diverse patient populations are expected to provide insights into the diagnostic and prognostic value of FOXP4-AS1.
Funding source: The National Nature Science Foundation of China
Award Identifier / Grant number: U20A20343 and 82200673
-
Research ethics: The local Institutional Review Board deemed the study exempt from review.
-
Informed consent: Informed consent was obtained from all individuals included in this study.
-
Author contributions: All authors have accepted responsibility for the entire content of this manuscript and approved its submission.
-
Competing interests: Authors state no conflict of interest.
-
Research funding: This study was funded by the National Nature Science Foundation of China (82200673).
References
1. Durot, M, Bourguignon, PY, Schachter, V. Genome-scale models of bacterial metabolism: reconstruction and applications. FEMS Microbiol Rev 2009;33:164–90. https://doi.org/10.1111/j.1574-6976.2008.00146.x.Search in Google Scholar PubMed PubMed Central
2. Sherman, RM, Salzberg, SL. Pan-genomics in the human genome era. Nat Rev Genet 2020;21:243–54. https://doi.org/10.1038/s41576-020-0210-7.Search in Google Scholar PubMed PubMed Central
3. Kaye, AM, Wasserman, WW. The genome atlas: navigating a new era of reference genomes. Trends Genet 2021;37:807–18. https://doi.org/10.1016/j.tig.2020.12.002.Search in Google Scholar PubMed
4. Wang, K, Shi, X, Zhu, Z, Hao, X, Chen, L, Cheng, S, et al.. Mendelian randomization analysis of 37 clinical factors and coronary artery disease in East Asian and European populations. Genome Med 2022;14:63. https://doi.org/10.1186/s13073-022-01067-1.Search in Google Scholar PubMed PubMed Central
5. Aung, N, Vargas, JD, Yang, C, Fung, K, Sanghvi, MM, Piechnik, SK, et al.. Genome-wide association analysis reveals insights into the genetic architecture of right ventricular structure and function. Nat Genet 2022;54:783–91. https://doi.org/10.1038/s41588-022-01083-2.Search in Google Scholar PubMed
6. Grishin, D, Gusev, A. Allelic imbalance of chromatin accessibility in cancer identifies candidate causal risk variants and their mechanisms. Nat Genet 2022;54:837–49. https://doi.org/10.1038/s41588-022-01075-2.Search in Google Scholar PubMed PubMed Central
7. Derrien, T, Johnson, R, Bussotti, G, Tanzer, A, Djebali, S, Tilgner, H, et al.. The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome Res 2012;22:1775–89. https://doi.org/10.1101/gr.132159.111.Search in Google Scholar PubMed PubMed Central
8. Birney, E, Stamatoyannopoulos, JA, Dutta, A, Guigó, R, Gingeras, TR, Margulies, EH, et al.. Identification and analysis of functional elements in 1 % of the human genome by the ENCODE pilot project. Nature 2007;447:799–816. https://doi.org/10.1038/nature05874.Search in Google Scholar PubMed PubMed Central
9. Ponting, CP, Oliver, PL, Reik, W. Evolution and functions of long noncoding RNAs. Cell 2009;136:629–41. https://doi.org/10.1016/j.cell.2009.02.006.Search in Google Scholar PubMed
10. Cabili, MN, Trapnell, C, Goff, L, Koziol, M, Tazon-Vega, B, Regev, A, et al.. Integrative annotation of human large intergenic noncoding RNAs reveals global properties and specific subclasses. Gene Dev 2011;25:1915–27. https://doi.org/10.1101/gad.17446611.Search in Google Scholar PubMed PubMed Central
11. Charles Richard, JL, Eichhorn, PJA. Platforms for investigating LncRNA functions. SLAS Technol. 2018;23:493–506. https://doi.org/10.1177/2472630318780639.Search in Google Scholar PubMed PubMed Central
12. Chen, J, Zhang, J, Gao, Y, Li, Y, Feng, C, Song, C, et al.. LncSEA: a platform for long non-coding RNA related sets and enrichment analysis. Nucleic Acids Res 2021;49:D969–d80. https://doi.org/10.1093/nar/gkaa806.Search in Google Scholar PubMed PubMed Central
13. Clark, MB, Mattick, JS. Long noncoding RNAs in cell biology. Semin Cell Dev Biol 2011;22:366–76. https://doi.org/10.1016/j.semcdb.2011.01.001.Search in Google Scholar PubMed
14. Tam, C, Wong, JH, Tsui, SKW, Zuo, T, Chan, TF, Ng, TB. LncRNAs with miRNAs in regulation of gastric, liver, and colorectal cancers: updates in recent years. Appl Microbiol Biotechnol 2019;103:4649–77. https://doi.org/10.1007/s00253-019-09837-5.Search in Google Scholar PubMed
15. Voellenkle, C, Garcia-Manteiga, JM, Pedrotti, S, Perfetti, A, De Toma, I, Da Silva, D, et al.. Implication of Long noncoding RNAs in the endothelial cell response to hypoxia revealed by RNA-sequencing. Sci Rep 2016;6:24141. https://doi.org/10.1038/srep24141.Search in Google Scholar PubMed PubMed Central
16. Sun, J, Chen, G, Jing, Y, He, X, Dong, J, Zheng, J, et al.. LncRNA expression profile of human thoracic aortic dissection by high-throughput sequencing. Cell Physiol Biochem : Int J Exp Cell Physiol Biochem Pharmacol 2018;46:1027–41. https://doi.org/10.1159/000488834.Search in Google Scholar PubMed
17. Ma, Y, Zhang, J, Wen, L, Lin, A. Membrane-lipid associated lncRNA: a new regulator in cancer signaling. Cancer Lett 2018;419:27–9. https://doi.org/10.1016/j.canlet.2018.01.008.Search in Google Scholar PubMed
18. Qian, X, Zhao, J, Yeung, PY, Zhang, QC, Kwok, CK. Revealing lncRNA structures and interactions by sequencing-based approaches. Trends Biochem Sci 2019;44:33–52. https://doi.org/10.1016/j.tibs.2018.09.012.Search in Google Scholar PubMed
19. Zhu, J, Fu, H, Wu, Y, Zheng, X. Function of lncRNAs and approaches to lncRNA-protein interactions. Sci China Life Sci 2013;56:876–85. https://doi.org/10.1007/s11427-013-4553-6.Search in Google Scholar PubMed
20. Wang, X, Arai, S, Song, X, Reichart, D, Du, K, Pascual, G, et al.. Induced ncRNAs allosterically modify RNA-binding proteins in cis to inhibit transcription. Nature 2008;454:126–30. https://doi.org/10.1038/nature06992.Search in Google Scholar PubMed PubMed Central
21. Carlevaro-Fita, J, Rahim, A, Guigó, R, Vardy, LA, Johnson, R. Cytoplasmic long noncoding RNAs are frequently bound to and degraded at ribosomes in human cells. RNA 2016;22:867–82. https://doi.org/10.1261/rna.053561.115.Search in Google Scholar PubMed PubMed Central
22. Yang, Z, Jiang, S, Shang, J, Jiang, Y, Dai, Y, Xu, B, et al.. LncRNA: shedding light on mechanisms and opportunities in fibrosis and aging. Ageing Res Rev 2019;52:17–31. https://doi.org/10.1016/j.arr.2019.04.001.Search in Google Scholar PubMed
23. Yip, CW, Sivaraman, DM, Prabhu, AV, Shin, JW. Functional annotation of lncRNA in high-throughput screening. Essays Biochem 2021;65:761–73. https://doi.org/10.1042/ebc20200061.Search in Google Scholar
24. Tan, YT, Lin, JF, Li, T, Li, JJ, Xu, RH, Ju, HQ. LncRNA-mediated posttranslational modifications and reprogramming of energy metabolism in cancer. Cancer Commun 2021;41:109–20. https://doi.org/10.1002/cac2.12108.Search in Google Scholar PubMed PubMed Central
25. Zhong, L, Luo, Y, Fan, J. LncRNAs: promising therapeutic targets and biomarkers for ischemic stroke. Transl Stroke Res 2022. https://doi.org/10.1007/s12975-022-01048-x. [Epub ahead of print].Search in Google Scholar PubMed
26. Xu, Y, Zheng, Q, Zhou, T, Ye, B, Xu, Q, Meng, X. Necroptosis-related LncRNAs signature and subtypes for predicting prognosis and revealing the immune microenvironment in breast cancer. Front Oncol 2022;12:887318. https://doi.org/10.3389/fonc.2022.887318.Search in Google Scholar PubMed PubMed Central
27. Tang, S, Liao, K, Shi, Y, Tang, T, Cui, B, Huang, Z. Bioinformatics analysis of potential Key lncRNA-miRNA-mRNA molecules as prognostic markers and important ceRNA axes in gastric cancer. Am J Cancer Res 2022;12:2397–418.Search in Google Scholar
28. Huang, S, Li, D, Zhuang, L, Zhang, J, Wu, J. Identification of an epithelial-mesenchymal transition-related long non-coding RNA prognostic signature to determine the prognosis and drug treatment of hepatocellular carcinoma patients. Front Med 2022;9:850343. https://doi.org/10.3389/fmed.2022.850343.Search in Google Scholar PubMed PubMed Central
29. Cheng, Z, Jiang, S, Tao, R, Ge, H, Qin, J. Activating transcription factor 3-activated long noncoding RNA forkhead box P4-antisense RNA 1 aggravates colorectal cancer progression by regulating microRNA-423-5p/nucleus accumbens associated 1 axis. Bioengineered 2022;13:2114–29. https://doi.org/10.1080/21655979.2021.2023798.Search in Google Scholar PubMed PubMed Central
30. Shi, ZL, Zhou, GQ, Guo, J, Yang, XL, Yu, C, Shen, CL, et al.. Identification of a prognostic colorectal cancer model including LncRNA FOXP4-AS1 and LncRNA BBOX1-AS1 based on bioinformatics analysis. Cancer Biother Radiopharm 2022;37:893–906. https://doi.org/10.1089/cbr.2020.4242.Search in Google Scholar PubMed PubMed Central
31. Li, J, Lian, Y, Yan, C, Cai, Z, Ding, J, Ma, Z, et al.. Long non-coding RNA FOXP4-AS1 is an unfavourable prognostic factor and regulates proliferation and apoptosis in colorectal cancer. Cell Prolif 2017;50. https://doi.org/10.1111/cpr.12312.Search in Google Scholar PubMed PubMed Central
32. Niu, Y, Wang, G, Li, Y, Guo, W, Guo, Y, Dong, Z. LncRNA FOXP4-AS1 promotes the progression of esophageal squamous cell carcinoma by interacting with MLL2/H3K4me3 to upregulate FOXP4. Front Oncol 2021;11:773864. https://doi.org/10.3389/fonc.2021.773864.Search in Google Scholar PubMed PubMed Central
33. Li, Y, Li, T, Yang, Y, Kang, W, Dong, S, Cheng, S. YY1-induced upregulation of FOXP4-AS1 and FOXP4 promote the proliferation of esophageal squamous cell carcinoma cells. Cell Biol Int 2020;44:1447–57. https://doi.org/10.1002/cbin.11338.Search in Google Scholar PubMed
34. Yao, L, Wang, T, Wang, X. LncRNA FOXP4-AS1 serves as a biomarker for nasopharyngeal carcinoma diagnosis and prognosis. 3 Biotech 2021;11:25. https://doi.org/10.1007/s13205-020-02593-8.Search in Google Scholar PubMed PubMed Central
35. Zhong, LK, Zhou, J, He, X, He, BF, Zhou, XW, Zhu, JL, et al.. Long non-coding RNA FOXP4-AS1 acts as an adverse prognostic factor and regulates proliferation and apoptosis in nasopharyngeal carcinoma. Eur Rev Med Pharmacol Sci 2020;24:8008–16. https://doi.org/10.26355/eurrev_202008_22484.Search in Google Scholar PubMed
36. Yan, J, Zhou, Q. LncRNA FOXP4-AS1 silencing inhibits metastasis and epithelial-mesenchymal transition in nasopharyngeal carcinoma via miR-136-5p/MAPK1. Anticancer Drugs 2023. https://doi.org/10.1097/CAD.0000000000001510 [Epub ahead of print].Search in Google Scholar PubMed PubMed Central
37. Ye, J, Fu, Y, Wang, Z, Yu, J. Long non-coding RNA FOXP4-AS1 facilitates the biological functions of hepatocellular carcinoma cells via downregulating ZC3H12D by mediating H3K27me3 through recruitment of EZH2. Cell Biol Toxicol 2022;38:1047–62. https://doi.org/10.1007/s10565-021-09642-9.Search in Google Scholar PubMed PubMed Central
38. Liang, J, Wang, D, Qiu, G, Zhu, X, Liu, J, Li, H, et al.. Long noncoding RNA FOXP4-AS1 predicts unfavourable prognosis and regulates proliferation and invasion in hepatocellular carcinoma. BioMed Res Int 2021;2021:8850656. https://doi.org/10.1155/2021/8850656..Search in Google Scholar PubMed PubMed Central
39. Wang, D, Bai, T, Chen, G, Liu, J, Chen, M, Zhao, Y, et al.. Upregulation of long non-coding RNA FOXP4-AS1 and its regulatory network in hepatocellular carcinoma. OncoTargets Ther 2019;12:7025–38. https://doi.org/10.2147/ott.s220923.Search in Google Scholar
40. Binang, HB, Wang, YS, Tewara, MA, Du, L, Shi, S, Li, N, et al.. Expression levels and associations of five long non-coding RNAs in gastric cancer and their clinical significance. Oncol Lett 2020;19:2431–45. https://doi.org/10.3892/ol.2020.11311.Search in Google Scholar PubMed PubMed Central
41. Liao, C, Wang, A, Ma, Y, Liu, H. Long non-coding RNA FOXP4-AS1 is a prognostic biomarker and associated with immune infiltrates in ovarian serous cystadenocarcinoma. Medicine 2021;100:e27473. https://doi.org/10.1097/md.0000000000027473.Search in Google Scholar
42. Hua, T, Tian, YJ, Wang, RM, Zhao, CF, Kong, YH, Tian, RQ, et al.. FOXP4-AS1 is a favorable prognostic-related enhancer RNA in ovarian cancer. Biosci Rep 2021;41. https://doi.org/10.1042/bsr20204008..Search in Google Scholar
43. Zhao, J, Yang, T, Li, L. LncRNA FOXP4-AS1 is involved in cervical cancer progression via regulating miR-136-5p/CBX4 Axis. OncoTargets Ther 2020;13:2347–55. https://doi.org/10.2147/ott.s241818.Search in Google Scholar
44. Tao, HF, Shen, JX, Hou, ZW, Chen, SY, Su, YZ, Fang, JL. lncRNA FOXP4-AS1 predicts poor prognosis and accelerates the progression of mantle cell lymphoma through the miR-423-5p/NACC1 pathway. Oncol Rep 2021;45:469–80. https://doi.org/10.3892/or.2020.7897.Search in Google Scholar PubMed PubMed Central
45. Li, D, Li, Z, YanFei, W, Wang, Y, Shi, J, Liu, C, et al.. LncRNA FOXP4-AS promotes the progression of non-small cell lung cancer by regulating the miR-3184-5p/EIF5A axis. J Tissue Eng Regen Med 2022;16:335–45. https://doi.org/10.1002/term.3275.Search in Google Scholar PubMed PubMed Central
46. Xiong, J, Wu, L, Huang, L, Wu, C, Liu, Z, Deng, W, et al.. LncRNA FOXP4-AS1 promotes progression of Ewing sarcoma and is associated with immune infiltrates. Front Oncol 2021;11:718876. https://doi.org/10.3389/fonc.2021.718876.Search in Google Scholar PubMed PubMed Central
47. Yang, L, Ge, D, Chen, X, Qiu, J, Yin, Z, Zheng, S, et al.. FOXP4-AS1 participates in the development and progression of osteosarcoma by downregulating LATS1 via binding to LSD1 and EZH2. Biochem Biophys Res Commun 2018;502:493–500. https://doi.org/10.1016/j.bbrc.2018.05.198.Search in Google Scholar PubMed
48. Liu, XG, Xu, H, Chen, M, Tan, XY, Chen, XF, Yang, YG, et al.. Identify potential clinical significance of long noncoding RNA forkhead box P4 antisense RNA 1 in patients with early stage pancreatic ductal adenocarcinoma. Cancer Med 2020;9:2062–76. https://doi.org/10.1002/cam4.2818.Search in Google Scholar PubMed PubMed Central
49. Wu, X, Xiao, Y, Zhou, Y, Zhou, Z, Yan, W. LncRNA FOXP4-AS1 is activated by PAX5 and promotes the growth of prostate cancer by sequestering miR-3184-5p to upregulate FOXP4. Cell Death Dis 2019;10:472. https://doi.org/10.1038/s41419-019-1699-6.Search in Google Scholar PubMed PubMed Central
50. Huang, Y, Ling, A, Pareek, S, Huang, RS. Oncogene or tumor suppressor? Long noncoding RNAs role in patient’s prognosis varies depending on disease type. Transl Res : J Lab Clin Med 2021;230:98–110. https://doi.org/10.1016/j.trsl.2020.10.011.Search in Google Scholar PubMed PubMed Central
51. Nishida, N, Sugiyama, M, Kawai, Y, Naka, I, Iwamoto, N, Suzuki, T, et al.. Genetic association of IL17 and the importance of ABO blood group antigens in saliva to COVID-19. Sci Rep 2022;12:3854. https://doi.org/10.1038/s41598-022-07856-3.Search in Google Scholar PubMed PubMed Central
52. Wu, P, Ding, L, Li, X, Liu, S, Cheng, F, He, Q, et al.. Trans-ethnic genome-wide association study of severe COVID-19. Commun Biol 2021;4:1034. https://doi.org/10.1038/s42003-021-02549-5.Search in Google Scholar PubMed PubMed Central
53. Wu, S, Zhang, H, Liu, Y, Wang, R, Ye, S, Zeng, M, et al.. [Long non-coding RNAs show different expression profiles and display competing endogenous RNA potential in placenta accreta spectrum disorders]. Nanfang Yike Daxue Xuebao 2019;39:1253–9. https://doi.org/10.12122/j.issn.1673-4254.2019.10.19.Search in Google Scholar PubMed PubMed Central
54. Luo, X, Gao, Q, Zhou, T, Tang, R, Zhao, Y, Zhang, Q, et al.. FOXP4-AS1 inhibits papillary thyroid carcinoma proliferation and migration through the AKT signaling pathway. Front Oncol 2022;12:900836. https://doi.org/10.3389/fonc.2022.900836.Search in Google Scholar PubMed PubMed Central
55. Hanahan, D, Weinberg, RA. The hallmarks of cancer. Cell 2000;100:57–70. https://doi.org/10.1016/s0092-8674(00)81683-9.Search in Google Scholar PubMed
56. Martincorena, I, Campbell, PJ. Somatic mutation in cancer and normal cells. Science 2015;349:1483–9. https://doi.org/10.1126/science.aab4082.Search in Google Scholar PubMed
57. Zhang, B, Huang, J, Li, HL, Liu, T, Wang, YY, Waterman, P, et al.. GIDE is a mitochondrial E3 ubiquitin ligase that induces apoptosis and slows growth. Cell Res 2008;18:900–10. https://doi.org/10.1038/cr.2008.75.Search in Google Scholar PubMed PubMed Central
58. Earnshaw, WC, Martins, LM, Kaufmann, SH. Mammalian caspases: structure, activation, substrates, and functions during apoptosis. Annu Rev Biochem 1999;68:383–424. https://doi.org/10.1146/annurev.biochem.68.1.383.Search in Google Scholar PubMed
59. Chaffer, CL, Weinberg, RA. A perspective on cancer cell metastasis. Science 2011;331:1559–64. https://doi.org/10.1126/science.1203543.Search in Google Scholar PubMed
60. McGregor, AL, Hsia, CR, Lammerding, J. Squish and squeeze-the nucleus as a physical barrier during migration in confined environments. Curr Opin Cell Biol 2016;40:32–40. https://doi.org/10.1016/j.ceb.2016.01.011.Search in Google Scholar PubMed PubMed Central
61. Limia, CM, Sauzay, C, Urra, H, Hetz, C, Chevet, E, Avril, T. Emerging roles of the endoplasmic reticulum associated unfolded protein response in cancer cell migration and invasion. Cancers 2019;11. https://doi.org/10.3390/cancers11050631.Search in Google Scholar PubMed PubMed Central
62. Nguyen, DX, Massagué, J. Genetic determinants of cancer metastasis. Nat Rev Genet 2007;8:341–52. https://doi.org/10.1038/nrg2101.Search in Google Scholar PubMed
63. Rørth, P. Collective cell migration. Annu Rev Cell Dev Biol 2009;25:407–29.10.1146/annurev.cellbio.042308.113231Search in Google Scholar PubMed
64. Wang, W, Hu, M, Liu, H, Zhang, X, Li, H, Zhou, F, et al.. Global Burden of Disease Study 2019 suggests that metabolic risk factors are the leading drivers of the burden of ischemic heart disease. Cell Metab 2021;33:1943–56.e2. https://doi.org/10.1016/j.cmet.2021.08.005.Search in Google Scholar PubMed
65. Rafeek, RAM, Divarathna, MVM, Noordeen, F. A review on disease burden and epidemiology of childhood parainfluenza virus infections in Asian countries. Rev Med Virol 2021;31:e2164. https://doi.org/10.1002/rmv.2164.Search in Google Scholar PubMed
66. Karch, A. Modern burden of disease studies as a basis for decision-making processes in public health. Dtsch Arztebl Int 2021;118:135–6. https://doi.org/10.3238/arztebl.m2021.0152.Search in Google Scholar PubMed PubMed Central
67. Wang, F, Wang, C, Xia, H, Lin, Y, Zhang, D, Yin, P, et al.. Burden of prostate cancer in China, 1990-2019: findings from the 2019 global burden of disease study. Front Endocrinol 2022;13:853623. https://doi.org/10.3389/fendo.2022.853623.Search in Google Scholar PubMed PubMed Central
68. Dong, Y, Kang, H, Peng, R, Song, K, Guo, Q, Guan, H, et al.. Global, regional, and national burden of low bone mineral density from 1990 to 2019: results from the global burden of disease study 2019. Front Endocrinol 2022;13:870905. https://doi.org/10.3389/fendo.2022.870905.Search in Google Scholar PubMed PubMed Central
69. Onega, T, Barnhill, RL, Piepkorn, MW, Longton, GM, Elder, DE, Weinstock, MA, et al.. Accuracy of digital pathologic analysis vs traditional microscopy in the interpretation of melanocytic lesions. JAMA Dermatol 2018;154:1159–66. https://doi.org/10.1001/jamadermatol.2018.2388.Search in Google Scholar PubMed PubMed Central
70. Villéger, R, Lopès, A, Veziant, J, Gagnière, J, Barnich, N, Billard, E, et al.. Microbial markers in colorectal cancer detection and/or prognosis. World J Gastroenterol 2018;24:2327–47. https://doi.org/10.3748/wjg.v24.i22.2327.Search in Google Scholar PubMed PubMed Central
71. van Velzen, CL, Clur, SA, Rijlaarsdam, ME, Pajkrt, E, Bax, CJ, Hruda, J, et al.. Prenatal diagnosis of congenital heart defects: accuracy and discrepancies in a multicenter cohort. Ultrasound Obstet Gynecol 2016;47:616–22.https://doi.org/10.1002/uog.15742Search in Google Scholar PubMed
72. Wu, Y, Chen, H, Li, L, Zhang, L, Dai, K, Wen, T, et al.. Construction of novel gene signature-based predictive model for the diagnosis of acute myocardial infarction by combining random forest with artificial neural network. Front Cardiovasc Med 2022;9:876543. https://doi.org/10.3389/fcvm.2022.876543.Search in Google Scholar PubMed PubMed Central
73. Urdiales-Sánchez, S, González-Montaña, JR, Diaz-Pérez, R, Calvo-Calleja, P, Gutiérrez-Trueba, MA, Urdiales-Urdiales, J. Nodopathies in the early diagnosis of axonal forms of Guillain-Barré syndrome. Front Neurol 2022;13:902172. https://doi.org/10.3389/fneur.2022.902172.Search in Google Scholar PubMed PubMed Central
© 2023 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Review Articles
- Mitochondrial thermogenesis in cancer cells
- Application of indocyanine green in the management of oral cancer: a literature review
- Long non-coding RNA, FOXP4-AS1, acts as a novel biomarker of cancers
- The role of synthetic peptides derived from bovine lactoferricin against breast cancer cell lines: a mini-review
- Single cell RNA sequencing – a valuable tool for cancer immunotherapy: a mini review
- Research Articles
- Global patterns and temporal trends in ovarian cancer morbidity, mortality, and burden from 1990 to 2019
- The association between NRF2 transcriptional gene dysregulation and IDH mutation in Grade 4 astrocytoma
- More than just a KRAS inhibitor: DCAI abrogates the self-renewal of pancreatic cancer stem cells in vitro
- DUSP1 promotes pancreatic cancer cell proliferation and invasion by upregulating nephronectin expression
- IMMT promotes hepatocellular carcinoma formation via PI3K/AKT/mTOR pathway
- MiR-100-5p transfected MSCs-derived exosomes can suppress NSCLC progression via PI3K-AKT-mTOR
- Inhibitory function of CDK12i combined with WEE1i on castration-resistant prostate cancer cells in vitro and in vivo
- Prognostic potential of m7G-associated lncRNA signature in predicting bladder cancer response to immunotherapy and chemotherapy
- Case Reports
- A rare FBXO25–SEPT14 fusion in a patient with chronic myeloid leukemia treatment to tyrosine kinase inhibitors: a case report
- Stage I duodenal adenocarcinoma cured by a short treatment cycle of pembrolizumab: a case report
- Rapid Communication
- ROMO1 – a potential immunohistochemical prognostic marker for cancer development
- Article Commentary
- A commentary: Role of MTA1: a novel modulator reprogramming mitochondrial glucose metabolism
Articles in the same Issue
- Frontmatter
- Review Articles
- Mitochondrial thermogenesis in cancer cells
- Application of indocyanine green in the management of oral cancer: a literature review
- Long non-coding RNA, FOXP4-AS1, acts as a novel biomarker of cancers
- The role of synthetic peptides derived from bovine lactoferricin against breast cancer cell lines: a mini-review
- Single cell RNA sequencing – a valuable tool for cancer immunotherapy: a mini review
- Research Articles
- Global patterns and temporal trends in ovarian cancer morbidity, mortality, and burden from 1990 to 2019
- The association between NRF2 transcriptional gene dysregulation and IDH mutation in Grade 4 astrocytoma
- More than just a KRAS inhibitor: DCAI abrogates the self-renewal of pancreatic cancer stem cells in vitro
- DUSP1 promotes pancreatic cancer cell proliferation and invasion by upregulating nephronectin expression
- IMMT promotes hepatocellular carcinoma formation via PI3K/AKT/mTOR pathway
- MiR-100-5p transfected MSCs-derived exosomes can suppress NSCLC progression via PI3K-AKT-mTOR
- Inhibitory function of CDK12i combined with WEE1i on castration-resistant prostate cancer cells in vitro and in vivo
- Prognostic potential of m7G-associated lncRNA signature in predicting bladder cancer response to immunotherapy and chemotherapy
- Case Reports
- A rare FBXO25–SEPT14 fusion in a patient with chronic myeloid leukemia treatment to tyrosine kinase inhibitors: a case report
- Stage I duodenal adenocarcinoma cured by a short treatment cycle of pembrolizumab: a case report
- Rapid Communication
- ROMO1 – a potential immunohistochemical prognostic marker for cancer development
- Article Commentary
- A commentary: Role of MTA1: a novel modulator reprogramming mitochondrial glucose metabolism